ArticlePDF Available

The prosurvival Bcl-2 Homolog Bfl-1/A1 is a direct transcriptional target of NF-kB that blocks TNF-?? induced apoptosis

Authors:
  • Janssen Pharmaceutical Companies of Johnson & Johnson

Abstract and Figures

Bcl-2-family proteins are key regulators of the apoptotic response. Here, we demonstrate that the pro-survival Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target of NF-kappaB. We show that bfl-1 gene expression is dependent on NF-kappaB activity and that it can substitute for NF-kappaB to suppress TNFalpha-induced apoptosis. bfl-1 promoter analysis identified an NF-kappaB site responsible for its Rel/NF-kappaB-dependent induction. The expression of bfl-1 in immune tissues supports the protective role of NF-kappaB in the immune system. The activation of Bfl-1 may be the means by which NF-kappaB functions in oncogenesis and promotes cell resistance to anti-cancer therapy.
Content may be subject to copyright.
RESEARCH COMMUNICATION
The prosurvival Bcl-2 homolog
Bfl-1/A1 is a direct
transcriptional target of NF-B
that blocks TNF-induced
apoptosis
Wei-Xing Zong,
1,2
Leonard C. Edelstein,
1,2
Cailin Chen,
1
Judy Bash,
1,3,5
and Ce´line Ge´linas
1,4,6
1
Center for Advanced Biotechnology and Medicine,
2
Graduate
Program in Biochemistry and Molecular Biology, University
of Medicine and Dentistry of New Jersey–Robert Wood
Johnson Medical School (UMDNJ–RWJMS),
3
Graduate
Program in Microbiology and Molecular Genetics, Rutgers
University,
4
Department of Biochemistry, UMDNJ–RWJMS,
Piscataway New Jersey 08854-5638 USA
Bcl-2-family proteins are key regulators of the apoptotic
response. Here, we demonstrate that the pro-survival
Bcl-2 homolog Bfl-1/A1 is a direct transcriptional target
of NF-B. We show that bfl-1 gene expression is depen-
dent on NF-B activity and that it can substitute for
NF-B to suppress TNF-induced apoptosis. bfl-1 pro-
moter analysis identified an NF-B site responsible for
its Rel/NF-B-dependent induction. The expression of
bfl-1 in immune tissues supports the protective role of
NF-B in the immune system. The activation of Bfl-1
may be the means by which NF-B functions in onco-
genesis and promotes cell resistance to anti-cancer
therapy.
Received October 15, 1998; revised version accepted January
6, 1999.
Binding of the proinflammatory cytokine tumor necrosis
factor (TNF) to its receptor triggers competing signal-
ing pathways that determine whether a cell lives or dies.
Whereas one pathway is conducive to cell death, the
other leads to activation of Rel/NF-B transcription fac-
tors and the coincident inhibition of apoptosis (for re-
view, see Nagata 1997). Accumulating evidence supports
a proactive role for NF-B in the inhibition of cell death
induced by TNF and other death-causing agents (for
review, see Van Antwerp et al. 1998). Whereas the acti-
vation of NF-B blocks cell killing, its inhibition en-
hances the cytotoxicity of TNF and promotes apoptosis
in various cell systems, demonstrating the need for NF-
B function for cell survival (Beg et al. 1995; Beg and
Baltimore 1996; White et al. 1995; Liu et al. 1996; Van
Antwerp et al. 1996; Wang et al. 1996;Wu et al. 1996; Cai
et al. 1997; Zong et al. 1997). The protective effect of
NF-B is dependent on RNA and protein synthesis, sug-
gesting that it regulates the expression of genes that con-
fer resistance to death-inducing signals (for review, see
Nagata, 1997). The finding that an intact transactivation
domain is required for Rel/NF-B factors to block cell
death agrees with this hypothesis (Zong et al. 1998).
Proteins of the Bcl-2 family play critical roles in deter-
mining cell fate in the apoptotic pathway. Although
some members antagonize cell death, others exhibit a
proapoptotic activity (for reviews, see Reed 1996; Adams
and Cory 1998). By subtractive cDNA cloning, we iden-
tified the prosurvival Bcl-2-homolog Bfl-1/A1 as a direct
transcriptional target of NF-B. We show that ectopi-
cally expressed Rel proteins and stimuli that activate
endogenous NF-B factors up-regulate bfl-1 gene expres-
sion and that this is inhibited by a dominant IB␣⌬N
transgene. Expression of Bfl-1 alone conferred resistance
to TNF cytotoxicity, indicating that it can substitute
for NF-B to suppress apoptosis. bfl-1 promoter analysis
identified a consensus NF-B site responsible for its Rel-
dependent induction. Together, these results demon-
strate that NF-B directly activates Bfl-1/A1 to inhibit
programmed cell death. The preferential expression of
bfl-1 in immune tissues supports the protective role of
NF-B in the immune system.
Results and Discussion
We used a PCR-selected subtractive cDNA cloning ap-
proach to identify anti-apoptotic genes under Rel/NF-B
control. The HeLa-derived HtTA–CCR43 cell line,
which conditionally expresses c-Rel under the control of
a tetracycline-regulated promoter, is resistant to TNF-
induced cell death upon induction of c-Rel (Bash et al.
1997; Zong et al. 1998). mRNA from HtTA–CCR43 cells
conditionally expressing c-Rel was reverse transcribed
and subjected to subtraction and PCR amplification. A
subtracted cDNA fragment of 700 bp was found to be
identical to bfl-1, a member of the Bcl-2 family of apop-
tosis inhibitors. Bfl-1 was originally isolated from fetal
liver and from cytokine-treated endothelial cells (Choi et
al. 1995; Karsan et al. 1996) and shares 72% amino acid
identity with its mouse homolog A1 (Lin et al. 1993).
Northern blot analysis during a time course of c-Rel
induction confirmed the up-regulation of bfl-1 tran-
scripts in HtTA–CCR43 cells, with kinetics that paral-
leled those of c-rel and of the Rel/NF-B target gene i
b
(Fig. 1a). Induction of the transactivation-competent
RelA subunit of NF-B also led to a sharp increase in
bfl-1 mRNA levels in the tetracycline-regulated HtTA–
RelA cell line (Fig. 1b, lanes 4–6; Zong et al. 1998). In
contrast no expression was detected in response to the
p50/NF-B1 protein, which lacks a defined transcription
activation domain (Fig. 1b, lanes 1–3). Thus, bfl-1 gene
expression was specifically up-regulated upon ectopic
expression of transcriptionally active Rel/NF-B sub-
units.
[Key Words: Rel; NF-B; Bfl-1; A1; TNF; apoptosis]
5
Present address: Cancer Institute of New Jersey, New Brunswick, New
Jersey 08854 USA.
6
Corresponding author.
E-MAIL gelinas@mbcl.rutgers.edu; FAX (732) 235-5289.
382 GENES & DEVELOPMENT 13:382–387 © 1999 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/99 $5.00; www.genesdev.org
Next, we verified the ability of endogenous NF-B fac-
tors to activate bfl-1 gene expression in response to vari-
ous stimuli and in different cells. TNFstrongly induced
bfl-1 transcripts in human HT1080 fibrosarcoma cells
(Fig. 2a, lanes 1,2). bfl-1 mRNA levels were also strongly
elevated in human Jurkat T-cells stimulated with phor-
bol 12-myristate 13-acetate (PMA) plus ionomycin (lanes
3,4). Likewise, the treatment of mouse 70Z/3 pre-B cells
with bacterial lipopolysaccharides (LPS) promoted the
accumulation of bfl-1/a1 transcripts (lanes 5,6). Consis-
tent with the nuclear NF-B activity found in mouse
WEHI-231 B cells (Liou et al. 1994; Miyamoto et al.
1994), basal bfl-1/a1 expression was observed in these
cells (lanes 7,9,11). bfl-1/a1 mRNAs were further in-
duced by different NF-B-inducing agents, including
TNF, LPS, or PMA (lanes 8,10,12).
The activity of NF-B is controlled by its association
with IB factors in the cytoplasm. In contrast to wild-
type IB, which undergoes proteasome-mediated degra-
dation in response to stimuli, an amino-terminally de-
leted IB transgene is resistant to signal-induced degra-
dation and acts as a constitutive repressor of NF-Bin
Jurkat–IB␣⌬N cells (Chu et al. 1997). We investigated
whether NF-B was necessary for the stimuli-induced
activation of bfl-1 by characterizing the effects of wild-
type IB and a trans-dominant IB␣⌬N inhibitor on
bfl-1 gene expression. The levels of 28S rRNA and of
mRNA for the NF-B-regulated gene interleukin-8 (IL-8)
were monitored as controls. Similar to the induction of
bfl-1 in the parental Jurkat T-cell line (Fig. 2a, lanes 3,4),
bfl-1 transcripts were sharply elevated by PMA plus
ionomycin treatment of T cells expressing a wild-type
IBtransgene (Fig. 2b, lanes 1,2). In contrast, the induc-
tion of bfl-1 was reduced by 83% and that of IL-8 was
decreased by 65% in cells expressing IB␣⌬N (lanes 3,4).
This indicated that nuclear NF-B activity is important
for bfl-1 gene expression.
Similar to c-rel, relA, and their target i
b
, bfl-1 was
found to be abundantly expressed in discrete immune
tissues. A Northern blot survey showed high levels of
bfl-1 transcripts in human spleen, lymph nodes, periph-
eral blood leukocytes and bone marrow (Fig. 3, lanes
6,7,9,10). Little expression was seen in thymus, fetal
liver, ovary, and small intestine, whereas none was de-
tected in prostate, testis, or colon (lanes 1–5,8,11). These
results suggest a physiological role for bfl-1 in promoting
the survival of defined cell lineages in the immune sys-
tem.
In agreement with the anti-apoptotic activity of NF-B
toward TNF, the transient cotransfection of a CMV-
bfl-1 expression vector with a CMV–-galactosidase re-
porter plasmid significantly suppressed the TNF-in-
duced killing of human HeLa cells in the presence of the
protein synthesis inhibitor cycloheximide (CHX; Fig.
4a). Quantitation of cell survival showed that the tran-
sient expression of bfl-1 increased the viability of HeLa
cells 8.5-fold in comparison to cells transfected with the
control pCMV plasmid (Fig. 4b). Cell protection was also
observed in the human HtTA-1 and HT1080 cell lines
(Fig. 4b). Moreover, Bfl-1 also suppressed TNF-induced
apoptosis under conditions in which endogenous NF-B
activity was inhibited by a serine-to-alanine mutant of
Figure 1. bfl-1 gene expression is activated by c-Rel and RelA
but not by p50/NF-B1. (a) Expression of bfl-1 transcripts in
HtTA–CCR43 cells maintained in the presence (lane 1)orab-
sence of tetracycline for 12, 24, 36, or 48 hr to induce c-rel
expression (lanes 2–5). The blot was successively hybridized to
32
P-labeled probes for bfl-1, i
b
,c-rel, and gapdh.(b) bfl-1 gene
expression in HtTA–p50 and HtTA–RelA cells maintained in
the presence (lanes 1,4) or absence of tetracycline for 24 or 48 hr
to induce the expression of p50 or relA (lanes 2,3,5,6). The blot
was hybridized to bfl-1 and actin probes.
Figure 2. The expression of bfl-1 is dependent on endogenous
Rel/NF-B activity. (a) The activation of NF-B induces bfl-1
gene expression. Human HT1080 cells untreated or treated with
TNF(lanes 1,2), human Jurkat T cells treated with DMSO as a
control (lane 3) or with PMA plus ionomycin (lane 4), mouse
70Z/3 pre-B cells untreated or treated with LPS (lanes 5,6),
mouse WEHI-231 B cells untreated or treated with TNF(lanes
7,8), LPS (lanes 9,10), or PMA (lanes 11,12). Total RNA (20 µg)
was hybridized to bfl-1 and actin probes. (b) The induction of
bfl-1 gene expression is repressed by a dominant IB␣⌬N inhibi-
tor. Jurkat T cells expressing wild-type IB or an IB␣⌬N trans-
gene were treated with DMSO alone (lanes 1,3)orwithPMA
plus ionomycin (lanes 2,4). Total RNA (20 µg) was hybridized to
32
P-labeled probes for bfl-1, il-8, and 28S rRNA.
Bfl-1/A1 is under Rel/NF-B control
GENES & DEVELOPMENT 383
IB that is resistant to signal-induced degradation
(IBM; Van Antwerp et al. 1996). Whereas HeLa cells
transfected with IBM were sensitized to TNF alone,
the cotransfection of Bfl-1 rescued the cells from cytoly-
sis (Fig. 4c). These results indicated that Bfl-1 can sub-
stitute for NF-B to block TNF-induced cytolysis.
These data agree with the protective effect of Bfl-1 and
its homolog A1 toward various inducers of cell death
(D’Sa-Eipper et al. 1996; Karsan et al. 1996; Lin et al.
1996). Together, our findings support a role for Bfl-1 as a
survival factor in the NF-B-signaling pathway that con-
fers resistance to TNF-induced apoptosis.
Immunofluorescence studies localized Bfl-1 to the cy-
toplasm (data not shown). This is compatible with the
subcellular localization of its homolog Bcl-2 (Krajewski
et al. 1993). Consistent with this observation, the inhibi-
tory activity of Bfl-1 toward TNF-induced cell death
was similar to that of Bcl-2 in HeLa cells. Whereas Bfl-1
enabled 45%–50% of the cells to survive treatment
with TNF plus CHX, Bcl-2 enabled 50%–60% of the
cells to escape cytolysis in transient transfection assays
(data not shown). However, further studies are needed to
determine whether Bfl-1 utilizes the same mechanisms
as Bcl-2 to inhibit cell death.
Analysis of the bfl-1 promoter region identified se-
quence elements responsible for its Rel-dependent in-
duction. Nested PCR amplification of adaptor-ligated
human genomic DNA libraries generated products of
1.4, 1.3, 0.4, and 0.2 kb (GenomeWalker-kit, Clontech).
All shared a common 3 end derived from the 5 end of
the bfl-1 cDNA and extended 5 into adjacent genomic
sequences. The purified products were directionally
cloned into a promoterless reporter plasmid for analysis
(Fig. 5a). The detailed characterization of the promoter
will be described elsewhere (L.C. Edelstein and C.
Ge´linas, in prep.).
bfl-1 promoter activity was assayed by transient trans-
fection of HeLa cells in the presence or absence of a
CMV-c-rel vector. An IL6BCAT reporter plasmid con-
taining three B DNA sites served as a positive control.
As shown in Figure 5b, all four clones showed minimal
basal activity on their own. The cotransfection of
pCMV–c-rel enhanced expression from the −1374/+81
and −1240/+81 promoter constructs by 12- and 9-fold,
respectively. In contrast, the activity of the −367/+81
and −129/+81 constructs was only marginally increased
by c-rel. Our mapping of a consensus NF-B DNA site at
position −833 relative to the transcription start site of
bfl-1 agreed with these results (GGGGATTTACC; Fig.
5a). Consistent with these findings, cell treatment with
a physiological inducer of NF-B also activated the bfl-1
promoter. As shown in Figure 5c, TNFstimulated CAT
expression from the bfl-1 promoter, similar to its effect
on the control IL6BCAT reporter plasmid. Inactivation
of the consensus NF-B motif in the context of the bfl-1
promoter region provided direct evidence that bfl-1 is
under Rel/NF-B control (GTTT
ATTTACC; −1374/
+81mB). Mutation of this NF-B site decreased gene
expression significantly in the presence of c-Rel (Fig. 5d).
Figure 3. bfl-1 is highly expressed in human immune tissues
and correlates with endogenous Rel/NF-B activity. Multiple
tissue Northern blots (Clontech) human II (lanes 15) and hu-
man immune system II (lanes 611) were successively hybrid-
ized to
32
P-labeled probes for bfl-1, i
b
,c-rel, relA, and actin.
Figure 4. Bfl-1 suppresses TNF-induced cell death. (a) HeLa
cells were cotransfected with pCMV–
-gal, together with an
empty pCMV vector or pCMV–bfl-1. The cells were treated
with CHX alone or together with TNF for 16 hr, stained with
X-gal, and photographed. (b) Quantitation of cell survival upon
expression of bfl-1. The viability of HeLa, HtTA-1, and HT1080
cells transfected as described in a represents the ratio of cells
expressing gal in wells treated with TNF plus CHX over that
in wells treated with CHX alone. Cells from a minimum of 10
randomly chosen fields were counted. The average survival
from three independent experiments is shown. (c) HeLa cells
were cotransfected with pCMV-
-gal and an empty pCMV vec-
tor or pCMV–IBM, alone or together with pCMV-bfl-1. The
cells were treated with TNF alone for 16 hr and stained with
X-gal. Cell survival represents the ratio of cells expressing
-gal
in wells treated with TNF over that in wells left untreated.
The average survival from three experiments is shown.
Zong et al.
384 GENES & DEVELOPMENT
Together, these findings demonstrate that bfl-1 is a pro-
survival gene under direct Rel/NF-B control.
Prosurvival members of the Bcl-2 family have been
shown to block apoptosis in lymphoid cells under con-
ditions in which NF-B activity was inhibited (Wu et al.
1996; for review, see Sonenshein 1997). This raised the
possibility that some members of the Bcl-2 family may
lie downstream of NF-B in the survival cascade. Our
demonstration that bfl-1/a1 is a transcriptional target of
NF-B provides the first direct evidence that a Bcl-2-fam-
ily member is controlled by NF-B proteins. These find-
ings are consistent with previous reports indicating that
bfl-1/a1 gene expression is induced by proinflammatory
cytokines in endothelial, leukemic, and hemopoietic
cells (Moreb and Schweder 1997; Lin et al. 1993; Karsan
et al. 1996b).
Our data also show clearly that bfl-1 can suppress
TNF-induced cytolysis. The anti-apoptotic activity of
Bfl-1 in this context agrees with work indicating that
Bfl-1/A1 can confer resistance to a variety of death in-
ducers in different cells (D’Sa-Eipper et al. 1996; Karsan
et al. 1996a; Lin et al. 1996). Thus, Bfl-1
may be viewed as an important player
in the survival pathway. It remains
possible that bfl-1 may act in combina-
tion with other anti-apoptotic genes to
block cell death efficiently in response
to different stimuli and in different
cells. For example, whereas NF-Bwas
recently implicated in inducing expres-
sion of the death inhibitors c-IAP1, c-
IAP2, and the TRAF1 (TNFR-associ-
ated factor 1) and TRAF2 factors, all
four proteins must act in combination
to efficiently block TNF-induced apop-
tosis in cells where NF-B is inactive
(Chu et al. 1997; Wang et al. 1998; You
et al. 1997). The presence of an NF-B
site in the promoter region of the zinc
finger protein A20 suggests that it may
also be under NF-B control (Krikos et
al. 1992), although A20 failed to rescue
RelA
−/−
cells from TNF-induced cy-
tolysis (Beg and Baltimore 1996). Simi-
larly, the immediate-early response
gene IEX-1L was shown to be involved
in NF-B-mediated cell survival, but its
mechanism of action remains to be
clarified (Wu et al. 1998). Preliminary
data from our laboratory agree with
these reports and suggest that other
anti-apoptotic factors are also regulated
by Rel/NF-B (C. Chen and C. Ge´linas,
in prep.). It will thus be important to
evaluate how their activities are coor-
dinated by the NF-B-signaling path-
way and to determine whether they
function individually or cooperatively
in response to different stimuli and in
different cellular environments.
The coinciding expression of bfl-1 and c-rel in the
white pulp of the spleen, the germinal centers of lym-
phatic tissues, and inflammatory cells (Carrasco et al.
1994; Jung-ha et al. 1998) supports a model whereby Bfl-1
may be a critical factor for carrying out the protective
role of Rel/NF-B in the immune system and during the
inflammatory response. Bfl-1 was shown previously to
cooperate with the adenovirus E1A protein in inducing
cell transformation and to be overexpressed in certain
cancers (Choi et al. 1995; D’Sa-Eipper et al. 1996). Al-
though the participation of Bfl-1 in oncogenesis is still a
topic of controversy (Jung-ha et al. 1998), the activation
of Bfl-1 by NF-B may also be a means by which NF-B
functions in oncogenesis and promotes the resistance of
tumor cells to anti-cancer therapy.
Materials and methods
Cells and endogenous NF-
B activation
Parental HtTA-1 cells and the HtTA-1-derived HtTA–CCR43, HtTA–
RelA, and HtTA–p50 cell clones that expressed c-rel, relA, and p50,re-
spectively, under tetracycline-regulated control have been described
Figure 5. The human bfl-1 promoter contains a consensus NF-B site responsible for
its Rel-dependent induction. (a) Schematic representation of CAT reporter gene con-
structs driven by various regions of the human bfl-1 promoter. (b) c-Rel-dependent
transactivation of the bfl-1 promoter. HtTA-1 cells were cotransfected with bfl-1–CAT
reporter plasmids together with pCMV–c-rel or an empty pCMV vector as a control.
The IL6BCAT plasmid containing three NF-B DNA sites derived from the IL6 pro-
moter was used as a positive control. The average CAT activity from three independent
experiments is shown. (c) TNF-inducible activation of the bfl-1 promoter. HtTA-1
cells were transfected with −1374/+81 bfl-1–CAT or the control IL6BCAT reporter
plasmid. Where indicated, cells were stimulated with TNF for 6 hr prior to harvest.
The average fold activation from three independent experiments is shown. (d) B site-
dependent activation of the bfl-1 promoter. Cells were cotransfected with wild-type
−1374/+81 bfl-1–CAT or the mutant −1374/+81mB–CAT reporter plasmid, with a
mutated NF-B site at position −833 (GTTTATTTACC), together with pCMV–c-rel or
an empty CMV vector as a control. IL6BCAT was used as a control.
Bfl-1/A1 is under Rel/NF-B control
GENES & DEVELOPMENT 385
(Gossen and Bujard 1992; Bash et al. 1997; Zong et al. 1998). Human HeLa
cervical carcinoma cells, HT1080 fibrosarcoma cells and Jurkat T-lym-
phocytic leukemia cells, and mouse 70Z/3 pre-B cells and WEHI-231
mature B-cells were obtained from ATCC. Jurkat–IBwt and Jurkat–
IB␣⌬N T cells were a gift of D.W. Ballard (Chu et al. 1997). Endogenous
NF-B activity was induced by treatment with TNF (Sigma; 10 ng/ml)
for 2 hr (WEHI-231) or 3 hr (HT1080), with PMA (50 ng per ml) plus
ionomycin (1 µ
M in DMSO) for 2 hr or DMSO alone as a control (0.05%),
with LPS (10 µg/ml) for 4 hr, or with PMA (100 n
M)for2hr.
Subtractive hybridization and cloning of a bfl-1 cDNA
Poly(A)
+
RNA was isolated from HtTA–CCR43 cells using a QuickPrep
RNA purification kit (Pharmacia). Purified mRNA (2 µg) was reverse
transcribed and subjected to subtractive hybridization with a PCR-Select
cDNA subtraction kit (Clontech). Subtracted cDNA fragments were
cloned in the pCRII vector (Invitrogen) and sequenced (Sequenase 2.0;
U.S. Biochemical). A full-length HA-tagged bfl-1 cDNA clone was ob-
tained by RT–PCR of total RNA from human HeLa cells in two succes-
sive rounds of amplification. The 5 primer used in the first round was
GCGTTCCAGATTACGCTAGCTTGATGACAGACTGTGAATTTG-
GA, and the 3 primer was CTGCTTAAGAGCTCTCAACATGATT-
GCTTCAGG. In the second round, the 5 primer containing an HA
tag was GGATCCGCCATGGCATACCCATATGATGTTCCAGATTA-
CGCT. The 3bfl-1-specific primer was identical to that used in the first
round. The amplification product was cloned in a pCMV vector for tran-
sient transfection assays (pcDNA3; Invitrogen). The bfl-1 cDNA se-
quence was confirmed with a T7 sequencing kit (Pharmacia).
Northern blot analysis
Total RNA (20 µg) extracted with RNAzol B (TEL-TEST) was fraction-
ated in a 1% agarose–formaldehyde gel and transferred onto a Hybond-
NX membrane (Amersham). The membrane was baked for 10 min at
80°C under vacuum and UV cross-linked with a Stratalinker (Stratagene).
Multiple tissue Northern blots were purchased from Clontech (human,
human II, human immune system II). Probes were generated by random
priming with Klenow polymerase in the presence of [
32
P]dCTP and
[
32
P] dGTP (Feinberg and Vogelstein 1983). Membranes were hybridized
in SSC (0.75
M NaCl, 75 mM Na citrate at pH 7.0), Denhardt’s
solution, 0.5% SDS, and 100 µg/ml sheared salmon sperm DNA at 65°C
overnight. Membranes were washed twice in SSC, 0.1% SDS, and
twice in 1× SSC, 0.1% SDS, at 65°C, followed by autoradiography.
TNF
-induced apoptosis
Cell resistance to TNF-induced apoptosis was assayed as described
(White et al. 1992). Cells (3 × 10
6
) were incubated with pCMV–
-gal
(5µg), together with pCMV–bfl-1 (15 µg) or an empty pCMV vector as a
control, and electroporated at 220 V, 960 µF using a Bio-Rad Gene Pulser.
The cells were then distributed equally into two 35-mm wells. After 24
hr, the cells were treated with CHX alone (30 µg/ml) or together with
TNF (10 ng/ml) for 16 hr. The cells were fixed and stained with X-gal
and photographed at a magnification of 200x. In assays of cell death
performed in the absence of CHX, HeLa cells were coelectroporated with
pCMV–
-gal (3 µg), an empty CMV vector, or pCMV–IBM (12 µg) to
constitutively repress NF-B, alone or together with pCMV–bfl-1 (6 µg).
Cells were then distributed equally into two 35-mm wells and treated 24
hr later with TNF (10 ng/ml) for 16 hr.
Cloning of the human bfl-1 promoter, transient CAT assays,
and mutagenesis
The human bfl-1 promoter region was isolated by nested PCR amplifi-
cation with a GenomeWalker-PromoterFinder kit (Clontech) and cloned
in a promoterless vector expressing a CAT reporter gene (pCAT-basic;
Promega). bfl-1 promoter activity was analyzed by transient transfection
of HtTA-1 cells with bfl-1–CAT reporter plasmids (3 µg) in the presence
of a CMV–c-rel expression vector (1 µg; Xu et al. 1993) or an empty pCMV
vector as a control. Where indicated, cells were treated with TNF (10
ng/ml) for 6 hr before harvest. An IL6BCAT reporter plasmid containing
three NF-B DNA sites from the IL6 promoter was used as a positive
control (Xu et al. 1993). The −1374/+81 bfl-1 promoter region cloned in
pAlter-1 was subjected to site-directed mutagenesis to inactivate the
consensus NF-B motif at position −833 (GTTT
ATTTACC, −1374/
+81mB; Altered Sites Mutagenesis System, Promega). Mutation of the
consensus NF-B site was confirmed by sequencing.
Acknowledgments
We are very grateful to C. Labrie for allowing C.C. to clone HA-bfl-1 in
his laboratory, to D.W. Ballard for Jurkat–IBwt and Jurkat–IB␣⌬N
cells, and to H. Bujard for the gift of HtTA-1 cells. We thank A. Rabson,
B. Rayet, A. Shatkin, and E. White for helpful comments on the manu-
script. This work was supported by grants from the National Institutes of
Health (NIH CA54999), The Council for Tobacco Research USA (4175),
and by the New Jersey Commission on Science and Technology. L.C.E. is
supported by NIH Biotechnology pre-doctoral training grant GM08339.
C.C. is a postdoctoral fellow of the New Jersey Commission on Cancer
Research and The Foundation of UMDNJ.
The publication costs of this article were defrayed in part by payment
of page charges. This article must therefore be hereby marked ‘advertise-
ment’ in accordance with 18 USC section 1734 solely to indicate this
fact.
References
Adams, J.M. and S. Cory. 1998. The Bcl-2 protein family: Arbiters of cell
survival. Science 281:1322–1326.
Bash, J., W.-X. Zong, and C. Ge´linas. 1997. c-Rel arrests the proliferation
of HeLa cells and affects critical regulators of the G1/S-phase tran-
sition. Mol. Cell. Biol. 17: 6526–6536.
Beg, A.A. and D. Baltimore. 1996. An essential role for NF-B in prevent-
ing TNF-induced cell death. Science 274: 782–784.
Beg, A.A., W.C. Sha, R.T. Bronson, and D. Baltimore. 1995. Constitutive
NF-B activation, enhanced granulopoiesis, and neonatal lethality in
IB-deficient mice. Genes & Dev. 9: 2736–2746.
Cai, Z., M. Korner, N. Tarantino, and S. Chouaib. 1997. IBoverexpres-
sion in human breast carcinoma MCF7 cells inhibits nuclear factor-
B activation but not tumor necrosis factor -induced apoptosis. J.
Biol. Chem. 272: 96–101.
Carrasco, D., F. Weih, and R. Bravo. 1994. Developmental expression of
the mouse c-rel proto-oncogene in hematopoietic organs. Develop-
ment 120: 2991–3004.
Choi, S.S., I.-C. Park, J.W. Yun, Y.C. Sung, S.I. Hong, and H.-S. Shin.
1995. A novel Bcl-2 related gene, Bfl-1, is overexpressed in stomach
cancer and preferentially expressed in bone marrow. Oncogene
11: 1693–1698.
Chu, Z.-L., T.A. McKinsey, L. Liu, J.J. Gentry, M.H. Malim, and D.W.
Ballard. 1997. Suppression of tumor necrosis factor-induced cell
death by inhibitor of apoptosis c-IAP2 is under NF-B control. Proc.
Natl. Acad. Sci. 94: 10057–10062.
D’Sa-Eipper, C., T. Subramanian, and G. Chinnadurai. 1996. Bfl-1, a Bcl-2
homologue, suppresses p53-induced apoptosis and exhibits potent co-
operative transforming activity. Cancer Res. 56: 3879–3882.
Feinberg, A.P. and B. Vogelstein. 1983. A technique for radiolabeling
DNA restriction endonuclease fragments to high specific activity.
Anal. Biochem. 132: 6–13.
Gossen, M. and H. Bujard. 1992. Tight control of gene expression in
mammalian cells by tetracycline-responsive promoters. Proc. Natl.
Acad. Sci. 89: 5547–5551.
Jung-ha, H., D. Kim, S.-B. Lee, S.-I. Hone, S.-Y. Park, J. Huh, C.-W. Kim,
S.-S. Kim, Y. Lee, S.S. Choi, and H.-S. Shin. 1998. Expression of Bfl-1
in normal and tumor tissues: Bfl-1 overexpression in cancer is attrib-
utable to its preferential expression in infiltrating inflammatory
cells. Hum. Pathol. 29: 723–728.
Karsan, A., E. Yee, and J.M. Harlan. 1996a. Endothelial cell death induced
by tumor necrosis factor is inhibited by the Bcl-2 family member,
A1. J. Biol. Chem. 271: 27201–27204.
Karsan, A., E. Yee, K. Kaushansky, and J.M. Harlan. 1996b. Cloning of a
human Bcl-2 homologue: Inflammatory cytokines induce human A1
in cultured endothelial cells. Blood 87: 3089–3096.
Krajewski, S., S. Tanaka, S. Takayama, M.J. Schibler, W. Fenton, and J.C.
Reed. 1993. Investigation of the subcellular distribution of the bcl-2
oncoprotein: Residence in the nuclear envelope, endoplasmic reticu-
lum, and outer mitochondrial membranes. Cancer Res. 53: 4701–
4714.
Krikos, A., C.D. Laherty, and V.M. Dixit. 1992. Transcriptional activa-
tion of the tumor necrosis factor -inducible zinc finger protein, A20,
is mediated by B elements. J. Biol. Chem. 267: 17971–17976.
Lin, E.Y., A. Orlofsky, M.S. Berger, and M.B. Prystowsky. 1993. Charac-
Zong et al.
386 GENES & DEVELOPMENT
terization of A1, a novel hemopoietic-specific early-response gene
with sequence similarity to bcl-2. J. Immunol. 151: 1979–1988.
Lin, E.Y., A. Orlofsky, H.G. Wang, J.C. Reed, and M.B. Prystowsky. 1996.
A1, a Bcl-2 family member, prolongs cell survival and permits my-
eloid differentiation. Blood 87: 983–992.
Liou, H.C., W.C. Sha, M.L. Scott, and D. Baltimore. 1994. Sequential
induction of NF-B/Rel family proteins during B-cell terminal differ-
entiation. Mol. Cell. Biol. 14: 5349–5359.
Liu, Z., H. Hsu, D.V. Goeddel, and M. Karin. 1996. Dissection of TNF
receptor 1 effector functions: JNK activation is not linked to apopto-
sis while NF-B activation prevents cell death. Cell 87: 565–576.
Miyamoto, S., M.J. Schmitt, and I.M. Verma. 1994. Qualitative changes
in the subunit composition of B-binding complexes during murine
B-cell differentiation. Proc. Natl. Acad. Sci. 91: 5056–5060.
Moreb, J.S. and M. Schweder. 1997. Human A1, a Bcl-2-related gene, is
induced in leukemic cells by cytokines as well as differentiating fac-
tors. Leukemia 11: 998–1004.
Nagata, S. 1997. Apoptosis by death factor. Cell 88: 355–365.
Reed, J.C. 1996. Mechanisms of Bcl-2 family protein function and dys-
function in health and disease. Behring Inst. Mitt. 97: 72–100.
Sonenshein, G.E. 1997. Rel/NF-B transcription factors and the control
of apoptosis. Sem. Cancer Biol. 8: 113–119.
Van Antwerp, D.J., S.J. Martin, T. Kafri, D.R. Green, and I.M. Verma.
1996. Suppression of TNF-induced apoptosis by NF-B. Science
274: 787–789.
Van Antwerp, D.J., S.J. Martin, I.M. Verma, and D.R. Green. 1998. Inhi-
bition of TNF-induced apoptosis by NF-B. Trends Cell Biol. 8 107–
111.
Wang, C.-Y., M.W. Mayo, R.G. Korneluk, D.V. Goeddel, and A.S. Bald-
win, Jr. 1998. NF-B antiapoptosis: induction of TRAF1 and TRAF2
and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science
281: 1680–1683.
Wang, C.-Y., M.W. Mayo, and A.S. Baldwin, Jr. 1996. TNF and cancer
therapy-induced apoptosis: potentiation by inhibition of NF-B. Sci-
ence 274: 784–787.
White, D.W., A. Roy, and T.D. Gilmore. 1995. The v-Rel oncoprotein
blocks apoptosis and proteolysis of IB in transformed chicken
spleen cells. Oncogene 10: 857–868.
White, E., P. Sabbatini, M. Debbas, W.S.M. Wold, D.I. Kusher, and L.R.
Gooding. 1992. The 19-kilodalton adenovirus E1B transforming pro-
tein inhibits programmed cell death and prevents cytolysis by tumor
necrosis factor . Mol. Cell. Biol. 12: 2570–2580.
Wu, M., H. Lee, R.E. Bellas, S.L. Schauer, M. Arsura, D. Katz, M.J.
FitzGerald, T.L. Rothstein, D.H. Sherr, and G.E. Sonenshein. 1996.
Inhibition of NF-B/Rel induces apoptosis of murine B cells. EMBO
J. 15: 4682–4690.
Wu, M.X., Z. Ao, K.V.S. Prasad, R. Wu, and S.F. Schlossman. 1998. IEX-
1L, an apoptosis inhibitor involved in NF-B-mediated cell survival.
Science 281: 998–1001.
Xu, X., C. Prorock, H. Ishikawa, E. Maldonado, Y. Ito, and C. Ge´linas.
1993. Functional interaction of the v-Rel and c-Rel oncoproteins with
the TATA-binding protein and association with transcription factor
IIB. Mol. Cell. Biol. 13: 6733–6741.
You, M., P.-T. Ku, R. Hrdlickova, and H.R. Bose. 1997. ch-IAP1, a mem-
ber of the inhibitor-of-apoptosis protein family, is a mediator of the
antiapoptotic activity of the v-Rel oncoprotein. Mol. Cell. Biol.
17: 7328–7341.
Zong, W.-X., M. Farrell, J. Bash, and C. Ge´linas. 1997. v-Rel prevents
apoptosis in transformed lymphoid cells and blocks TNF-induced
cell death. Oncogene 15: 971–980.
Zong, W.-X., J. Bash, and C. Ge´linas. 1998. Rel blocks both anti-Fas- and
TNF-induced apoptosis and an intact Rel transactivation domain is
essential for this effect. Cell Death Differ. 5: 963–972.
Bfl-1/A1 is under Rel/NF-B control
GENES & DEVELOPMENT 387
... Moreover, the ER can also mediate gene expression by coordinating with different transcription factors, such as Activating Protein 1 (AP1), Specificity Protein 1 (SP1), and Nuclear Factor Kappa B (NF-κB) [14]. Among the five antiapoptotic genes, three (BCL2, BCL2A1, and MCL1) can be regulated by NF-κB in different types of cancer cells [15][16][17]. ...
... NF-κB consists of five subunit proteins, p65 (RelA), RelB, c-Rel, p50 (NF-κB1), and p52 (NF-κB2), which can form homoand heterodimers to regulate gene expression [15][16][17]. Whether NF-κB can also mediate the expression of the five antiapoptotic genes in breast cancer cells is unclear. Moreover, a study has shown that BCL2 mRNA and protein levels are upregulated, while BCL2L1 mRNA and protein levels are downregulated, by E2 in the ER-positive MCF7 cell line. ...
... The changes in BCL2, BCL2A1, BCL2L2, and MCL1 mRNA levels in E2-treated HME1, T47D, and MCF7 cells suggested that these genes might be regulated at the transcriptional level. Several studies have shown that BCL2, BCL2A1, and MCL1 are the target genes of the NF-κB transcription factor [15][16][17]. We found that the promoters of the BCL2, BCL2A1, BCL2L2, and MCL1 genes, but not the promoter of the BCL2L1 gene, contained an NF-κB binding site when we predicted the promoters using the consensus DNA sequence of 5ʹ-GGGRNYYYCC-3ʹ in which R represented a purine, Y represented a pyrimidine, and N represented any nucleotide ( Fig. 2A). ...
Article
Full-text available
Circumvention of apoptosis by the elevation of antiapoptotic proteins is an important cause of carcinogenesis. The induction of antiapoptotic genes, including B-cell CLL/lymphoma 2 (BCL2), BCL2 related protein A1 (BCL2A1), BCL2 like 1 (BCL2L1), BCL2L2, and myeloid cell leukemia 1 (MCL1), has been observed in multiple cancers, including breast cancer. However, the underlying mechanisms of their overexpression are still being investigated. Here, we revealed that BCL2, BCL2A1, BCL2L2, and MCL1 but not BCL2L1 were overexpressed in estrogen receptor (ER)-positive breast cancer cells and clinical biopsies. Stimulation with estrogen in ER-positive cell lines resulted in a dose-dependent increase in BCL2, BCL2A1, BCL2L2, and MCL1 mRNA levels. Molecular investigation revealed that nuclear factor kappa B (NF-κB) recruited histone acetyltransferase p300 and nuclear receptor coactivator 3 (NCOA3) to form a transcriptional complex. This complex docked the promoters of BCL2, BCL2A1, BCL2L2, and MCL1 and activated their expression. Interestingly, estrogen exposure dose-dependently activated NCOA3. Depletion of the NCOA3-p300-NF-κB components or blockage of NCOA3 function with inhibitors (gossypol and bufalin) in ER-positive cells suppressed BCL2, BCL2A1, BCL2L2, and MCL1 expression, while also decreasing cell viability, colony formation, cell invasion, and tumor growth. Collectively, our results demonstrate an upstream signaling that activates four antiapoptotic genes in ER-positive breast cancer cells. Importantly, our results also imply that targeting NCOA3 or blocking the assembly of the NCOA3-p300-NF-κB complex may be promising therapeutic strategies for treating ER-positive breast cancer.
... For example, p53 induces apoptosis by upregulating the transcription of mRNAs encoding proapoptotic proteins such as BAX, PUMA, and Noxa [14,15] but repressing that of the mRNA encoding the antiapoptotic protein BCL-2 [13]. NF-κB and STAT3 drive the transcription of BCL-2 [16] and BFL1 [17] in the immune system to promote cell survival, thus maintaining immune homeostasis. Therefore, identifying a new transcription factor that responds to hyperglycaemia and activates the transcription of key players in the apoptotic process is clearly essential in understanding hyperglycaemia-induced tissue injury. ...
Article
Full-text available
Chronic hyperglycaemia is a devastating factor that causes diabetes-induced damage to the retina and kidney. However, the precise mechanism by which hyperglycaemia drives apoptotic cell death is incompletely known. Herein, we found that FOXD1, a FOX family transcription factor specifically expressed in the retina and kidney, regulated the transcription of BCL-2, a master regulator of cell survival. Intriguingly, the protein level of FOXD1, which responded negatively to hyperglycaemic conditions, was controlled by the TRIM21-mediated K48-linked polyubiquitination and subsequent proteasomal degradation. The TRIM21-FOXD1-BCL-2 signalling axis was notably active during diabetes-induced damage to murine retinal and renal tissues. Furthermore, we found that tartary buckwheat flavonoids effectively reversed the downregulation of FOXD1 protein expression and thus restored BCL-2 expression and facilitated the survival of retinal and renal tissues. In summary, we identified a transcription factor responsible for BCL-2 expression, a signalling axis (TRM21-FOXD1-BCL-2) underlying hyperglycaemia-triggered apoptosis, and a potential treatment for deleterious diabetic complications.
... MnSOD Scavenging of superoxide radicals Wong et al. (1989), Wan et al. (1994) Ferritin heavy chain Sequestration of iron resulting in reduced TNF-induced ROS accumulation Kwak et al. (1995), Pham et al. (2004) A20 a Stabilizes M1 ubiquitination in TNFR1 signaling complex Krikos et al. (1992), He and Ting (2002) Inhibits intrinsic apoptosis pathway Zong et al. (1999), Cheng et al. (2000) Bcl2 ...
Article
Full-text available
Tumor necrosis factor (TNF) receptor 1 (TNFR1), TNFR2 and fibroblast growth factor-inducible 14 (Fn14) belong to the TNF receptor superfamily (TNFRSF). From a structural point of view, TNFR1 is a prototypic death domain (DD)-containing receptor. In contrast to other prominent death receptors, such as CD95/Fas and the two TRAIL death receptors DR4 and DR5, however, liganded TNFR1 does not instruct the formation of a plasma membrane-associated death inducing signaling complex converting procaspase-8 into highly active mature heterotetrameric caspase-8 molecules. Instead, liganded TNFR1 recruits the DD-containing cytoplasmic signaling proteins TRADD and RIPK1 and empowers these proteins to trigger cell death signaling by cytosolic complexes after their release from the TNFR1 signaling complex. The activity and quality (apoptosis versus necroptosis) of TNF-induced cell death signaling is controlled by caspase-8, the caspase-8 regulatory FLIP proteins, TRAF2, RIPK1 and the RIPK1-ubiquitinating E3 ligases cIAP1 and cIAP2. TNFR2 and Fn14 efficiently recruit TRAF2 along with the TRAF2 binding partners cIAP1 and cIAP2 and can thereby limit the availability of these molecules for other TRAF2/cIAP1/2-utilizing proteins including TNFR1. Accordingly, at the cellular level engagement of TNFR2 or Fn14 inhibits TNFR1-induced RIPK1-mediated effects reaching from activation of the classical NFκB pathway to induction of apoptosis and necroptosis. In this review, we summarize the effects of TNFR2- and Fn14-mediated depletion of TRAF2 and the cIAP1/2 on TNFR1 signaling at the molecular level and discuss the consequences this has in vivo.
... BCL2A1 is an NF-κB target gene with an important role in the innate immune response. BCL2A1 expression was shown to depend on NF-κB activity, and can substitute for NF-κB to suppress tumor necrosis factor-α-induced apoptosis [36]. Previous studies suggested that activation of NF-κB inhibited infected epithelial cell apoptosis and facilitated parasite survival [37]. ...
Article
Full-text available
Background Cryptosporidium is second only to rotavirus as a cause of moderate-to-severe diarrhea in young children. There are currently no fully effective drug treatments or vaccines for cryptosporidiosis. MicroRNAs (miRNAs) are involved in regulating the innate immune response to Cryptosporidium parvum infection. In this study, we investigated the role and mechanism of miR-3976 in regulating HCT-8 cell apoptosis induced by C. parvum infection. Methods Expression levels of miR-3976 and C. parvum burden were estimated using real-time quantitative polymerase chain reaction (RT-qPCR) and cell apoptosis was detected by flow cytometry. The interaction between miR-3976 and B-cell lymphoma 2-related protein A1 (BCL2A1) was studied by luciferase reporter assay, RT-qPCR, and western blotting. Results Expression levels of miR-3976 were decreased at 8 and 12 h post-infection (hpi) but increased at 24 and 48 hpi. Upregulation of miR-3976 promoted cell apoptosis and inhibited the parasite burden in HCT-8 cells after C. parvum infection. Luciferase reporter assay indicated that BCL2A1 was a target gene of miR-3976. Co-transfection with miR-3976 and a BCL2A1 overexpression vector revealed that miR-3976 targeted BCL2A1 and suppressed cell apoptosis and promoted the parasite burden in HCT-8 cells. Conclusions The present data indicated that miR-3976 regulated cell apoptosis and parasite burden in HCT-8 cells by targeting BCL2A1 following C. parvum infection. Future study should determine the role of miR-3976 in hosts’ anti-C. parvum immunity in vivo. Graphical Abstract
... The transcription of multiple apoptosis-related genes has been found to be controlled by NF-κB [35][36][37][38] . NF-κB regulates the expression of two antiapoptotic members (BCL-XL and A1/Bfl-1) [39][40][41][42][43][44][45] , on the other hand, NF-κB2 suppresses the expression of the proapoptotic Bax protein 46 . The purpose of this research is to find the correlation between BCL2 and P52 in malignant cells of breast carcinoma. ...
... Consequently, CXCL1 activates its receptor CXCR2, triggering the activation of NF-κB [201]. This transcription factor directly increases the expression of anti-apoptotic Bcl-2 family proteins such as Bcl-2 [201], Bfl-1/A1 [202], and Bcl-xL [203], which inhibit apoptosis induced by chemotherapeutic agents. Additionally, CXCL1 recruits G-MDSC [52,57], which contribute to chemoresistance by secreting S100A8/9 [28]. ...
Article
Full-text available
C-X-C motif chemokine ligand 1 (CXCL1) is a member of the CXC chemokine subfamily and a ligand for CXCR2. Its main function in the immune system is the chemoattraction of neutrophils. However, there is a lack of comprehensive reviews summarizing the significance of CXCL1 in cancer processes. To fill this gap, this work describes the clinical significance and participation of CXCL1 in cancer processes in the most important reproductive cancers: breast cancer, cervical cancer, endometrial cancer, ovarian cancer, and prostate cancer. The focus is on both clinical aspects and the significance of CXCL1 in molecular cancer processes. We describe the association of CXCL1 with clinical features of tumors, including prognosis, ER, PR and HER2 status, and TNM stage. We present the molecular contribution of CXCL1 to chemoresistance and radioresistance in selected tumors and its influence on the proliferation, migration, and invasion of tumor cells. Additionally, we present the impact of CXCL1 on the microenvironment of reproductive cancers, including its effect on angiogenesis, recruitment, and function of cancer-associated cells (macrophages, neutrophils, MDSC, and Treg). The article concludes by summarizing the significance of introducing drugs targeting CXCL1. This paper also discusses the significance of ACKR1/DARC in reproductive cancers.
Article
Myosin heavy chain‐perinatal (MyHC‐perinatal) is one of two development‐specific myosin heavy chains expressed exclusively during skeletal muscle development and regeneration. The specific functions of MyHC‐perinatal are unclear, although mutations are known to lead to contracture syndromes such as Trismus‐pseudocamptodactyly syndrome. Here, we characterize the functions of MyHC‐perinatal during skeletal muscle differentiation and regeneration. Loss of MyHC‐perinatal function leads to enhanced differentiation characterized by increased expression of myogenic regulatory factors and differentiation index as well as reduced reserve cell numbers in vitro . Proteomic analysis revealed that loss of MyHC‐perinatal function results in a switch from oxidative to glycolytic metabolism in myofibers, suggesting a shift from slow type I to fast type IIb fiber type, also supported by reduced mitochondrial numbers. Paracrine signals mediate the effect of loss of MyHC‐perinatal function on myogenic differentiation, possibly mediated by non‐apoptotic caspase‐3 signaling along with enhanced levels of the pro‐survival apoptosis regulator Bcl2 and nuclear factor kappa‐B (NF‐κB). Knockdown of MyHC‐perinatal during muscle regeneration in vivo results in increased expression of the differentiation marker myogenin (MyoG) and impaired differentiation, evidenced by smaller myofibers, elevated fibrosis and reduction in the number of satellite cells. Thus, we find that MyHC‐perinatal is a crucial regulator of myogenic differentiation, myofiber oxidative phenotype and regeneration.
Article
Full-text available
Chronic myeloid leukemia (CML) was considered for a long time one of the most hostile leukemia that was incurable for most of the patients, predominantly due to the extreme resistance to chemotherapy. Part of the resistance to cell death (apoptosis) is the result of increased levels of anti-apoptotic and decreased levels of pro-apoptotic member of the BCL-2 family induced by the BCR-ABL1 oncoprotein. BCR-ABL1 is a constitutively active tyrosine kinase responsible for initiating multiple and oncogenic signaling pathways. With the development of specific BCR-ABL1 tyrosine kinase inhibitors (TKIs) CML became a much more tractable disease. Nevertheless, TKIs do not cure CML patients and a substantial number of them develop intolerance or become resistant to the treatment. Therefore, novel anti-cancer strategies must be developed to treat CML patients independently or in combination with TKIs. Here, we will discuss the mechanisms of BCR-ABL1-dependent and -independent resistance to TKIs and the use of BH3-mimetics as a potential tool to fight CML.
Article
Activated B cell-like diffuse large B-cell lymphoma (ABC-DLBCL) is the most aggressive form of DLBCL, with a significantly inferior prognosis due to resistance to the standard R-CHOP immunochemotherapy. Survival of ABC-DLBCL cells addicted to the constitutive activations of both canonical and noncanonical NF-κB signaling makes them attractive therapeutic targets. However, a pharmaceutical approach simultaneously targeting the canonical and noncanonical NF-κB pathway in the ABC-DLBCL cell is still lacking. Peptide-conjugated gold nanoclusters (AuNCs) have emerged unique intrinsic biomedical activities and possess a great potential in cancer theranostics. Here, we demonstrated a Au25 nanocluster conjugated by cell-penetrating peptides that can selectively repress the growth of ABC-DLBCL cells by inducing efficient apoptosis, more efficiently than glutathione (GSH)-conjugated AuNCs. The mechanism study showed that the cell-penetrating peptides enhanced the cellular internalization efficiency of AuNCs, and the selective repression in ABC-DLBCL cells is due to the inhibition of inherent constitutive canonical and noncanonical NF-κB activities by AuNCs. Several NF-κB target genes involved in chemotherapy resistance in ABC-DLBCL cells, including anti-apoptotic Bcl-2 family members and DNA damage repair proteins, were effectively down-regulated by the AuNC. The emerged novel activity of AuNCs in targeting both arms of NF-κB signaling in ABC-DLBCL cells may provide a promising candidate and a new insight into the rational design of peptide-conjugated Au nanomedicine for molecular targeting treatment of refractory lymphomas.
Article
Full-text available
A20 was first identified as a tumor necrosis factor (TNF) primary response transcript encoding a 790-amino acid protein with a unique zinc finger motif. Recently, A20 was shown to protect cells from TNF-induced cytotoxicity in a variety of cell lines. Nuclear run-on studies previously established that TNF activates A20 at the transcriptional level. To further characterize the mechanism by which TNF activates the A20 gene, we have cloned the A20 5'-flanking sequences and identified TNF-responsive elements within the promoter. The transcription initiation site was mapped by both primer extension and S1 nuclease protection experiments to a position 4.2 kilobases kB upstream of the initiator methionine; the first and second exon were separated by a 3.9-kb intron. Sequences upstream of the transcription start site were 76% GC-rich and contained six Sp1 binding sites and a TATA-like sequence at -29 but lacked a consensus CCAAT site. Transfection of Jurkat T-cells with an array of A20 promoter CAT constructs showed that two kappa-B elements residing at -54 and -66 were required for induction by TNF. Supporting this notion, DNA electrophoretic mobility shift assays using nuclear extracts from unstimulated and TNF-stimulated Jurkat cells demonstrated kappa-B-specific binding of a TNF-activated factor to an end-labeled probe containing the two A20 kappa-B sequences. Finally, evidence obtained from cotransfection experiments showed that A20 negatively regulated its own expression.
Article
Full-text available
The adenovirus E1A and E1B proteins are required for transformation of primary rodent cells. When expressed in the absence of the 19,000-dalton (19K) E1B protein, however, the E1A proteins are acutely cytotoxic and induce host cell chromosomal DNA fragmentation and cytolysis, analogous to cells undergoing programmed cell death (apoptosis). E1A alone can efficiently initiate the formation of foci which subsequently undergo abortive transformation whereby stimulation of cell growth is counteracted by continual cell death. Cell lines with an immortalized growth potential eventually arise with low frequency. Coexpression of the E1B 19K protein with E1A is sufficient to overcome abortive transformation to produce high-frequency transformation. Like E1A, the tumoricidal cytokine tumor necrosis factor alpha (TNF-alpha) evokes a programmed cell death response in many tumor cell lines by inducing DNA fragmentation and cytolysis. Expression of the E1B 19K protein by viral infection, by transient expression, or in transformed cells completely and specifically blocks this TNF-alpha-induced DNA fragmentation and cell death. Cosegregation of 19K protein transforming activity with protection from TNF-alpha-mediated cytolysis demonstrates that both activities are likely the consequence of the same function of the protein. Therefore, we propose that by suppressing an intrinsic cell death mechanism activated by TNF-alpha or E1A, the E1B 19K protein enhances the transforming activity of E1A and enables adenovirus to evade TNF-alpha-dependent immune surveillance.
Article
Full-text available
A20 was first identified as a tumor necrosis factor (TNF) primary response transcript encoding a 790-amino acid protein with a unique zinc finger motif. Recently, A20 was shown to protect cells from TNF-induced cytotoxicity in a variety of cell lines. Nuclear run-on studies previously established that TNF activates A20 at the transcriptional level. To further characterize the mechanism by which TNF activates the A20 gene, we have cloned the A20 5'-flanking sequences and identified TNF-responsive elements within the promoter. The transcription initiation site was mapped by both primer extension and S1 nuclease protection experiments to a position 4.2 kilobases (kb) upstream of the initiator methionine; the first and second exon were separated by a 3.9-kb intron. Sequences upstream of the transcription start site were 76% GC-rich and contained six Sp1 binding sites and a TATA-like sequence at -29 but lacked a consensus CCAAT site. Transfection of Jurkat T-cells with an array of A20 promoter CAT constructs showed that two kappa B elements residing at -54 and -66 were required for induction by TNF. Supporting this notion, DNA electrophoretic mobility shift assays using nuclear extracts from unstimulated and TNF-stimulated Jurkat cells demonstrated kappa B-specific binding of a TNF-activated factor to an end-labeled probe containing the two A20 kappa B sequences. Finally, evidence obtained from cotransfection experiments showed that A20 negatively regulated its own expression.
Article
Fas is a type I membrane protein belonging to the TNF receptor family. The Fas ligand (FasL) is a member of the TNF family, and induces apoptosis in Fas-bearing cells. The Fas-mediated apoptosis occurs in enucleated cells, and is executed by members of the ICE (interleukin-lβ-converting cn/ymc)/Ccd-3 protease family (caspases), and can be inhibited by Bcl2/Ced-9. The Fas cytoplasmic region carries a domain (a death domain) which is necessary and sufficient to mediate apoptosis. In the current model, the Fas engagement causes trimeriyation of the Fas death domain, which recruits a caspase (caspase 8) to the receptor via an adaptor molecule called FADD/ MORT 1. This seems to cause self-activation of the protease, and leads to the sequential activation of members of the caspase family. The Fas system is involved in peripheral clonal deletion, and in down-regulation of the immune reaction. Mouse mutations of lymphoproliferation (Ipr) and generalized lymphoproliferative disease (gld) which cause lymphadenopathy and splenomegaly, and accelerate autoimmune disease, are loss-of-function mutations in the Fas and FasL genes, respectively. FasL in activated T cells and NK (natural killer ) cells works as an effector of CTL and NK cells to remove the cells infected by virus, or cancerous cells. If mice were injected by the rccombinant FasL, the mice died with hours by the liver damage, suggesting that overfunction of this system causes disease such as hepatitis. In fact, the Fas ligand-neutralizing molecules efficiently prevented deelopment of hepatitis in mouse model systems. Using an ELISA system, we detected a significant amount of the soluble FasL in the serum of human patients who carry NK lymphoma or large granular lymphocytes leukemia, showing systemic tissue damage such as hepatitis and neutropenia.
Article
The NF-kappa B/Rel family of at least five transcription factor polypeptides is thought to function both as a developmental regulator in B cells and as a rapid response system in all cells. To examine this notion in more detail, we determined the protein contents of both the inducible and constitutive NF-kappa B/Rel activities in a pre-B-cell line, 70Z/3, and a mature B-cell line, WEHI 231. NF-kappa B p50/p65 is the major inducible nuclear complex after lipopolysaccharide or phorbol myristate acetate treatment of 70Z/3 cells. The constitutive and inducible complexes in WEHI 231 cells are mainly composed of p50 and Rel. The constitutive or induced activities are all sensitive to I kappa B-alpha, but this inhibitor is very short-lived in WEHI 231 cells, suggesting that the balance between synthesis and degradation of I kappa B-alpha determines whether a particular cell lineage has constitutive activity. A patterned expression of the NF-kappa B/Rel activator proteins emerges from an analysis of other B-lineage cell lines and splenic B cells: mainly p50 and p65 in pre-B (and non-B) cells, a predominance of Rel and p50 in mature B cells, and expression of p52 and RelB in plasmacytoma lines. This ordered pattern of regulators may reflect the requirement for expression of different genes during terminal B-cell differentiation because different combinations of NF-kappa B/Rel family members preferentially activate distinct kappa B sites in reporter constructs.
Article
Members of the NF-kappa B/Rel and inhibitor of apoptosis (IAP) protein families have been implicated in signal transduction programs that prevent cell death elicited by the cytokine tumor necrosis factor alpha (TNF). Although NF-kappa B appears to stimulate the expression of specific protective genes, neither the identities of these genes nor the precise role of IAP proteins in this anti-apoptotic process are known. We demonstrate here that NF-kappa B is required for TNF-mediated induction of the gene encoding human c-IAP2. When overexpressed in mammalian cells, c-IAP2 activates NF-kappa B and suppresses TNF cytotoxicity. Both of these c-IAP2 activities are blocked in vivo by coexpressing a dominant form of Ikappa B that is resistant to TNF-induced degradation. In contrast to wild-type c-IAP2, a mutant lacking the C-terminal RING domain inhibits NF-kappa B induction by TNF and enhances TNF killing. These findings suggest that c-IAP2 is critically involved in TNF signaling and exerts positive feedback control on NF-kappa B via an Ikappa B targeting mechanism. Functional coupling of NF-kappa B and c-IAP2 during the TNF response may provide a signal amplification loop that promotes cell survival rather than death.
Article
Control elements of the tetracycline-resistance operon encoded in Tn10 of Escherichia coli have been utilized to establish a highly efficient regulatory system in mammalian cells. By fusing the tet repressor with the activating domain of virion protein 16 of herpes simplex virus, a tetracycline-controlled transactivator (tTA) was generated that is constitutively expressed in HeLa cells. This transactivator stimulates transcription from a minimal promoter sequence derived from the human cytomegalovirus promoter IE combined with tet operator sequences. Upon integration of a luciferase gene controlled by a tTA-dependent promoter into a tTA-producing HeLa cell line, high levels of luciferase expression were monitored. These activities are sensitive to tetracycline. Depending on the concentration of the antibiotic in the culture medium (0-1 microgram/ml), the luciferase activity can be regulated over up to five orders of magnitude. Thus, the system not only allows differential control of the activity of an individual gene in mammalian cells but also is suitable for creation of "on/off" situations for such genes in a reversible way.
Article
A technique for conveniently radiolabeling DNA restriction endonuclease fragments to high specific activity is described. DNA fragments are purified from agarose gels directly by ethanol precipitation and are then denatured and labeled with the large fragment of DNA polymerase I, using random oligonucleotides as primers. Over 70% of the precursor triphosphate is routinely incorporated into complementary DNA, and specific activities of over 10(9) dpm/microgram of DNA can be obtained using relatively small amounts of precursor. These "oligolabeled" DNA fragments serve as efficient probes in filter hybridization experiments.
Article
Programmed cell death (apoptosis) is an active process which is genetically encoded and plays an important role in several cellular activities such as embryonic development, deletion of autoreactive T-cells and homeostasis. Several genes regulating apoptosis have been reported, including p53, one of the tumor suppressor genes, c-myc, one of the proto-oncogenes, and various kinds of Bcl-2 related genes. A new cDNA clone which is homologous to Bcl-2, named as Bfl-1 were isolated from a human fetal liver at 22 week of gestation. This clone was identified by computer analysis of random cDNA sequences that were obtained in an effort to expand the expressed sequence tag (EST) databases to be used for human genome analysis. The homology was recognized by 72% amino acid identity to the murine A1 gene, a member of the Bcl-2-related genes. The homology to the BH1 and BH2 domains of Bcl-2 was especially significant, suggesting that Bfl-1 is a new member of the Bcl-2-related genes. Bfl-1 is abundantly expressed in the bone marrow and at a low level in some other tissues. Interestingly, a correlation was noted between the expression level of Bfl-1 gene and the development of stomach cancer in eight sets of clinical samples. It is conceivable that Bfl-1 is involved in the promotion of the cell survival in the stomach cancer development or progression.